Skip to main content
Erschienen in: Investigational New Drugs 5/2019

04.12.2018 | PRECLINICAL STUDIES

A novel tetravalent bispecific antibody targeting programmed death 1 and tyrosine-protein kinase Met for treatment of gastric cancer

verfasst von: Weihua Hou, Qingyun Yuan, Xingxing Yuan, Yuxiong Wang, Wei Mo, Huijie Wang, Min Yu

Erschienen in: Investigational New Drugs | Ausgabe 5/2019

Einloggen, um Zugang zu erhalten

Summary

Background Redirecting T cells to tumor cells using bispecific antibodies (BsAbs) is emerging as a potent cancer therapy. The main concept of this strategy is to cross-link tumor cells and T cells by simultaneously binding to cell surface tumor-associated antigen (TAA) and the CD3ƹ chain. However, immune checkpoint programmed cell death ligand-1 (PD-L1) on tumor cells or other myeloid cells upreglulated remarkablely after the treatment of CD3-binding BsAbs, leads to the generation of suppressed microenvironment for immune evasion and tumor progression. Although this resistance could be partially reversed by anti-PD-L1 treatment, targeting two pathways through one antibody-based molecule may provide a strategic advantage over the combination of BsAbs and immune checkpoint inhibitors. Methods We developed two novel BsAbs PD-1/c-Met DVD-Ig and IgG-scFv both targeting PD-1 to restore the immune effector function of T cells and engaging them to tumor cells via binding to cellular-mesenchymal to epithelial transition factor (c-Met). Binding activities, T cell activation and proliferation were analyzed by flow cytometry. Cell Cytotoxicity and cytokine release were measured using LDH release assay and ELISA, respectively. Anti-tumor response in vivo was evaluated by generate xenograft models in NOD-SCID mice. Results These bispecific antibodies exhibited effective antitumor activity against high- and low- c-Met-expressing gastric cancer cell lines in vitro and mediated strong tumor growth inhibition in human gastric cancer xenograft models. Conclusion The engagement of the PD-1/PD-L1 blockade to c-Met-overexpressing cancer cells is a promising strategy for the treatment of gastric cancer and potentially other malignancies.
Literatur
5.
Zurück zum Zitat Lennerz JK, Kwak EL, Ackerman A, Michael M, Fox SB, Bergethon K, Lauwers GY, Christensen JG, Wilner KD, Haber DA, Salgia R, Bang YJ, Clark JW, Solomon BJ, Iafrate AJ (2011) MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J Clin Oncol Off J Am Soc Clin Oncol 29(36):4803–4810. https://doi.org/10.1200/JCO.2011.35.4928 CrossRef Lennerz JK, Kwak EL, Ackerman A, Michael M, Fox SB, Bergethon K, Lauwers GY, Christensen JG, Wilner KD, Haber DA, Salgia R, Bang YJ, Clark JW, Solomon BJ, Iafrate AJ (2011) MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J Clin Oncol Off J Am Soc Clin Oncol 29(36):4803–4810. https://​doi.​org/​10.​1200/​JCO.​2011.​35.​4928 CrossRef
9.
Zurück zum Zitat Dreier T, Lorenczewski G, Brandl C, Hoffmann P, Syring U, Hanakam F, Kufer P, Riethmuller G, Bargou R, Baeuerle PA (2002) Extremely potent, rapid and costimulation-independent cytotoxic T-cell response against lymphoma cells catalyzed by a single-chain bispecific antibody. Int J Cancer 100(6):690–697. https://doi.org/10.1002/ijc.10557 CrossRefPubMed Dreier T, Lorenczewski G, Brandl C, Hoffmann P, Syring U, Hanakam F, Kufer P, Riethmuller G, Bargou R, Baeuerle PA (2002) Extremely potent, rapid and costimulation-independent cytotoxic T-cell response against lymphoma cells catalyzed by a single-chain bispecific antibody. Int J Cancer 100(6):690–697. https://​doi.​org/​10.​1002/​ijc.​10557 CrossRefPubMed
13.
14.
Zurück zum Zitat Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, Noppeney R, Viardot A, Hess G, Schuler M, Einsele H, Brandl C, Wolf A, Kirchinger P, Klappers P, Schmidt M, Riethmuller G, Reinhardt C, Baeuerle PA, Kufer P (2008) Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321(5891):974–977. https://doi.org/10.1126/science.1158545 CrossRef Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, Noppeney R, Viardot A, Hess G, Schuler M, Einsele H, Brandl C, Wolf A, Kirchinger P, Klappers P, Schmidt M, Riethmuller G, Reinhardt C, Baeuerle PA, Kufer P (2008) Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321(5891):974–977. https://​doi.​org/​10.​1126/​science.​1158545 CrossRef
17.
Zurück zum Zitat Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, Chen S, Klein AP, Pardoll DM, Topalian SL, Chen L (2012) Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4(127):127ra137. https://doi.org/10.1126/scitranslmed.3003689 CrossRef Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, Chen S, Klein AP, Pardoll DM, Topalian SL, Chen L (2012) Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4(127):127ra137. https://​doi.​org/​10.​1126/​scitranslmed.​3003689 CrossRef
19.
Zurück zum Zitat Junttila TT, Li J, Johnston J, Hristopoulos M, Clark R, Ellerman D, Wang BE, Li Y, Mathieu M, Li G, Young J, Luis E, Lewis Phillips G, Stefanich E, Spiess C, Polson A, Irving B, Scheer JM, Junttila MR, Dennis MS, Kelley R, Totpal K, Ebens A (2014) Antitumor efficacy of a bispecific antibody that targets HER2 and activates T cells. Cancer Res 74(19):5561–5571. https://doi.org/10.1158/0008-5472.CAN-13-3622-T CrossRef Junttila TT, Li J, Johnston J, Hristopoulos M, Clark R, Ellerman D, Wang BE, Li Y, Mathieu M, Li G, Young J, Luis E, Lewis Phillips G, Stefanich E, Spiess C, Polson A, Irving B, Scheer JM, Junttila MR, Dennis MS, Kelley R, Totpal K, Ebens A (2014) Antitumor efficacy of a bispecific antibody that targets HER2 and activates T cells. Cancer Res 74(19):5561–5571. https://​doi.​org/​10.​1158/​0008-5472.​CAN-13-3622-T CrossRef
20.
Zurück zum Zitat Sun LL, Wang P, Clark R, Hristopoulos M, Ellerman D, Mathieu M, Chu Y-W, Wang H, Totpal K, Ebens AJ, Polson AG, Gould S (2016) Preclinical characterization of combinability and potential synergy of anti-CD20/CD3 T-cell dependent bispecific antibody with chemotherapy and PD-1/PD-L1 blockade. Blood 128(22):4168–4168CrossRef Sun LL, Wang P, Clark R, Hristopoulos M, Ellerman D, Mathieu M, Chu Y-W, Wang H, Totpal K, Ebens AJ, Polson AG, Gould S (2016) Preclinical characterization of combinability and potential synergy of anti-CD20/CD3 T-cell dependent bispecific antibody with chemotherapy and PD-1/PD-L1 blockade. Blood 128(22):4168–4168CrossRef
25.
Zurück zum Zitat Wu C, Ying H, Grinnell C, Bryant S, Miller R, Clabbers A, Bose S, McCarthy D, Zhu RR, Santora L, Davis-Taber R, Kunes Y, Fung E, Schwartz A, Sakorafas P, Gu J, Tarcsa E, Murtaza A, Ghayur T (2007) Simultaneous targeting of multiple disease mediators by a dual-variable-domain immunoglobulin. Nat Biotechnol 25(11):1290–1297. https://doi.org/10.1038/nbt1345 CrossRefPubMed Wu C, Ying H, Grinnell C, Bryant S, Miller R, Clabbers A, Bose S, McCarthy D, Zhu RR, Santora L, Davis-Taber R, Kunes Y, Fung E, Schwartz A, Sakorafas P, Gu J, Tarcsa E, Murtaza A, Ghayur T (2007) Simultaneous targeting of multiple disease mediators by a dual-variable-domain immunoglobulin. Nat Biotechnol 25(11):1290–1297. https://​doi.​org/​10.​1038/​nbt1345 CrossRefPubMed
41.
Zurück zum Zitat Michaud NR, Jani JP, Hillerman S, Tsaparikos KE, Barbacci-Tobin EG, Knauth E, Putz H Jr, Campbell M, Karam GA, Chrunyk B, Gebhard DF, Green LL, Xu JJ, Dunn MC, Coskran TM, Lapointe JM, Cohen BD, Coleman KG, Bedian V, Vincent P, Kajiji S, Steyn SJ, Borzillo GV, Los G (2012) Biochemical and pharmacological characterization of human c-Met neutralizing monoclonal antibody CE-355621. mAbs 4(6):710–723. https://doi.org/10.4161/mabs.22160 CrossRefPubMedPubMedCentral Michaud NR, Jani JP, Hillerman S, Tsaparikos KE, Barbacci-Tobin EG, Knauth E, Putz H Jr, Campbell M, Karam GA, Chrunyk B, Gebhard DF, Green LL, Xu JJ, Dunn MC, Coskran TM, Lapointe JM, Cohen BD, Coleman KG, Bedian V, Vincent P, Kajiji S, Steyn SJ, Borzillo GV, Los G (2012) Biochemical and pharmacological characterization of human c-Met neutralizing monoclonal antibody CE-355621. mAbs 4(6):710–723. https://​doi.​org/​10.​4161/​mabs.​22160 CrossRefPubMedPubMedCentral
Metadaten
Titel
A novel tetravalent bispecific antibody targeting programmed death 1 and tyrosine-protein kinase Met for treatment of gastric cancer
verfasst von
Weihua Hou
Qingyun Yuan
Xingxing Yuan
Yuxiong Wang
Wei Mo
Huijie Wang
Min Yu
Publikationsdatum
04.12.2018
Verlag
Springer US
Erschienen in
Investigational New Drugs / Ausgabe 5/2019
Print ISSN: 0167-6997
Elektronische ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-018-0689-3

Weitere Artikel der Ausgabe 5/2019

Investigational New Drugs 5/2019 Zur Ausgabe

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.